You are on page 1of 12

Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.

com
379

REVIEW

Genetics of congenital hypothyroidism This article is available free on JMG online


via the JMG Unlocked open access trial,
funded by the Joint Information Systems
Committee. For further information, see
S M Park, V K K Chatterjee http://jmg.bmjjournals.com/cgi/content/
full/42/2/97
...............................................................................................................................

J Med Genet 2005;42:379–389. doi: 10.1136/jmg.2004.024158

Congenital hypothyroidism is the most common neonatal In mammals the thyroid gland is the first
glandular tissue to appear in development,
metabolic disorder and results in severe arising from two regions of the endodermal
neurodevelopmental impairment and infertility if untreated. pharynx which migrate and associate to become
Congenital hypothyroidism is usually sporadic but up to 2% the characteristic bi-lobed structure.4 The median
anlage, which eventually forms the follicular
of thyroid dysgenesis is familial, and congenital cells, arises from the midline of the anterior
hypothyroidism caused by organification defects is often pharyngeal floor between the first and second
recessively inherited. The candidate genes associated with branchial arches, being visible from days 16 to 17
of human gestation. At the same time two lateral
this genetically heterogeneous disorder form two main anlagen (ultimobranchial bodies), which mainly
groups: those causing thyroid gland dysgenesis and those become the parafollicular calcitonin secreting C
causing dyshormonogenesis. Genes associated with cells but also contribute to a significant number
of follicular cells,5 develop as caudal projections
thyroid gland dysgenesis include the TSH receptor in non-
from the fourth or fifth pharyngeal pouches. The
syndromic congenital hypothyroidism, and Gsa and the thyroid gland begins to trap iodide and therefore
thyroid transcription factors (TTF-1, TTF-2, and Pax-8), to secrete thyroid hormones only at 10 to 12
associated with different complex syndromes that include weeks of human gestation,4 with the trans-
placental passage of maternal thyroid hor-
congenital hypothyroidism. Among those causing mones before this being important for fetal
dyshormonogenesis, the thyroid peroxidase and development.
thyroglobulin genes were initially described, and more The essential role of thyroid hormones in
central nervous system (CNS) maturation has
recently PDS (Pendred syndrome), NIS (sodium iodide been clearly demonstrated,6 and congenital
symporter), and THOX2 (thyroid oxidase 2) gene defects. hypothyroidism is eminently treatable by thyr-
There is also early evidence for a third group of congenital oxine replacement. Screening for neonatal
hypothyroidism has therefore been established
hypothyroid conditions associated with iodothyronine on a worldwide basis, beginning in Canada
transporter defects associated with severe neurological in 1974 and in the United Kingdom in 1982.
sequelae. This review focuses on the genetic aspects of The development of infants with congenital
hypothyroidism has been revolutionised with
primary congenital hypothyroidism. the institution of early and adequate treatment
........................................................................... afforded by screening, thereby preventing intel-
lectual impairment and infertility. This is wit-
nessed by the normal growth and neurological

C
ongenital hypothyroidism is detected at a
development in affected children, and by a
rate of 1 in 3000 to 4000 live births, making
dramatic increase in IQ, from an average of less
it the most common congenital endocrine
than 80 to the mean of the general population.7
disorder.1 On a worldwide basis, hypothyroidism,
Most cases of congenital hypothyroidism occur
including congenital forms, results most com-
sporadically. However, dyshormonogenetic cases
monly from iodine deficiency. Otherwise con-
are often recessively inherited, and recent cohort
genital thyroid gland insufficiency results from
analyses estimate that approximately 2% of cases
developmental abnormalities at any level of the
with thyroid dysgenesis are familial.8 It is also
See end of article for
hypothalamic-pituitary-thyroid axis. Congenital
noteworthy that congenital hypothyroidism is
authors’ affiliations hypothyroidism is most commonly caused by
....................... associated with an increased incidence of birth
defects in thyroid development leading to thyroid
defects, with surveys in the United Kingdom
Correspondence to:
dysgenesis (85%), which in turn consists of
reporting a non-thyroidal congenital anomaly
Dr S M Park, Department either thyroid agenesis (40% of all cases) or
rate of 7% and an increased prevalence of
of Clinical Genetics, Box failure of the gland to descend normally during
134, Addenbrooke’s chromosomal anomalies (1.5%).9 10
embryological development with or without
Hospital, Hills Road, The candidate genes associated with primary
Cambridge CB2 2QQ, UK;
ectopy (40%), or hypoplasia of a eutopic gland.2
congenital hypothyroidism can be divided into
soo-mi.park@ The remaining cases are associated with either a
addenbrookes.nhs.uk goitre or a normal thyroid gland.3 Rarely, central
(secondary) hypothyroidism may be caused by Abbreviations: ERSD, endoplasmic reticulum storage
Received in revised form disease; NIS, sodium iodide symporter; PHP,
pituitary or hypothalamic disease leading to
18 October 2004 pseudohypoparathyroidism; PPHP, pseudopseudo-
Accepted for publication deficiency of thyrotropin (TSH) or thyrotropin hypoparathyroidism; PTH, parathyroid hormone;
3 November 2004 releasing hormone (TRH), respectively, which TPO, thyroid peroxidase; TRH, thyrotropin releasing
....................... will not be covered by this review. hormone; TSH, thyroid stimulating hormone (thyrotropin)

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
380 Park, Chatterjee

two main groups: those causing thyroid gland dysgenesis and alterations in TSH signalling pathways could result in
those associated with defects in the organification of iodide, defective thyroid development. The hyt/hyt mouse is homo-
leading to dyshormonogenesis. Genes associated with thyroid zygous for a loss of function mutation in the fourth
gland dysgenesis include those causing non-syndromic transmembrane domain of the TSHR, resulting in hypothyr-
congenital hypothyroidism (TSH receptor) and those causing oidism and thyroid hypoplasia postnatally.16
syndromic congenital hypothyroidism (TITF-1, TITF-2, PAX-8, In contrast to mouse models, where an intact TSH-TSHR
and Gsa). Genes associated with dyshormonogenesis include signalling pathway does not appear to be a prerequisite for
those for thyroid peroxidase (TPO), thyroglobulin (TG), the the development of a normal sized thyroid gland in utero,17
sodium iodide symporter (NIS), pendrin (PDS), and most this pathway is clearly important for the development of a
recently, thyroid oxidase 2 (THOX2). Recent evidence normally sized fully differentiated gland in utero in
suggests a third group of congenital hypothyroid conditions humans.18 Homozygous or compound heterozygous muta-
associated with defects in the iodothyronine transporter, tions in the human TSHR gene resulting in TSH resistance
MCT8, where hypothyroidism is associated with severe were first described in 1995.19 Since then, additional loss of
neurological deficits. function TSHR mutations have been identified in other
In this review we will focus on the genetics of the primary families showing variable TSH resistance because of reduced
congenital hypothyroidism. We will summarise the different thyroid sensitivity to TSH (fig 1).20–32 In some of these
phenotypes associated with the currently known genetic families, compensated TSH resistance was associated with
defects and outline informative investigative management. euthyroid hyperthyrotropinaemia and either a normal or a
hypoplastic thyroid gland.19 20 27 29–31 Mild or borderline
THYROID DYSGENESIS AND NON-SYNDROMIC hypothyroidism, with greatly raised serum TSH associated
CONGENITAL HYPOTHYROIDISM with low normal or slightly low thyroid hormone levels and a
The TSH receptor gene (TSHR) encodes a transmembrane normal thyroid gland, was seen in two families.21 22 At the
receptor present on the surface of follicular cells which other end of the spectrum, severe hypothyroidism associated
mediates the effects of TSH secreted by the anterior pituitary with a hypoplastic thyroid gland or even apparent athyreosis
and is critical for the development and function of the has been reported.23–26 28 32
thyroid gland. It belongs to a subfamily of heptahelical G Inactivating mutations of the human TSHR are therefore
protein coupled receptors that have a common structure associated with three phenotypes:
consisting of seven transmembrane segments, three extra-
cellular and three intracellular loops, an extracellular amino N fully compensated TSH resistance to TSH;
terminal domain, and an intracytoplasmic carboxyl terminal N partially compensated TSH resistance to TSH;
tail (fig 1).11 The actions of TSH on the thyrocyte occur
principally by receptor mediated activation of Gsa and
N severe uncompensated resistance to TSH.

subsequent generation of intracellular cyclic adenosine In common with other G protein coupled receptors,
monophosphate (cAMP).12 The human TSHR gene is located activating mutations have also been described in the TSHR,
on chromosome 14q31 and the extracellular domain of the causing thyroid hyperfunctioning adenomas when somatic,33
receptor is encoded by nine exons, whereas the transmem- or non-autoimmune congenital hyperthyroidism with germ-
brane and intracellular portions are encoded by a single large line abnormalities.34 It is of note that heterozygosity for a
exon.13 14 TSHR mutation (W546X) is increasingly being reported in
The initiation of expression of the TSHR on day 14 of families from a white background,20 21 32 35 with a rate of
mouse embryogenesis, at the onset of thyroid differentia- approximately 0.6% in a cohort of euthyroid Welsh subjects.35
tion after completion of gland migration,15 suggests that These results have therefore raised the possibility that the

NH2 M R P A D L L Q L V L L L D L P R D L G GMG C S S P P C E C HQ E E D F R V T K D I Q R C
T
P
S
N Y F S H S E L Q Q L T V D L S V Y L R S L N P L N S F A H S P I T R L H T E I L K L T Q T S P P L
L
S
K
V T H L L I
R
N T R N L T Y L D P D A L K E L P L L K F L G L F N T G L KM F P D L T K V Y S T D
L
Intron 6 F
F

A
O L K T GN F A YGQ V S T F GNN Y L K L T L T E NC L GQ F AN V P I
S T MY P
ND T L E L I Intron 5/exon 6
V
Y
G C+3 AG AA
L N K N K Y L T V I D K D A F GG V Y S G P S L L D V S Q T S V T A L P S K G L E H L K E L I A R N
T
W
T
L
S OM S S E C L S E L L G R L K K QN K F A C C H S P Y S L D A R T L H L F S L S L P L K K L
R
O
R K S V NA L N S P L H QE Y E E N L G D S I V G V K E K S K F OD T HNNA H Y Y V F F E E O
E
D
E

D E G P NF E D S K P T C V M D E S D C C
I T Y D Y H S D F AQ L T E E Q P N K L E OG F G I I

M Y NH A L DW I C L PMD T
G
Y E
Y O
T V
S E
T
del (AC) 655
K S G K P
E T V S
P A I I
L L N
R T G
C
Y
S
A
L K
P S
K
Frameshift I V V L D V N T S T U A Y L N L L I I L

SV
F W
L I
A S
T F
G L
V
I V
F V
A L
S A
Y F
L V
I L A
L
V FA A L T L N Y F S P L N
L L E S L V I P L C S
NG CR S L P A MA N A
Membrane V F V
L
F D A V Y T W G C
V C I F P F L
L A F T L V C T D AY
L I NL I V I M I F F I
L T S T L F A C A V K L L V A T K A
H
V K
R W
B H
R L
Y M
R
F O R D
V
L N V F
Y L K L
L
R
A
I I R L
A L
S
T K N K K
F AM R L D R P
OY N P G
T F
D K D G
I
C
K
COOH L V T OM Y E E S I OGM K K P T L H S N E I L E Y V D F MN H L GO R M D H T V K O V O I D T S N K P P V R OG R Y AOAO R

Figure 1 Cartoon of the TSH receptor showing the positions of all the loss of function mutations reported to date. Missense mutations are shown in the
circles, frameshift and deletion mutations are indicated by arrows, and splice site mutations are marked.

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
Genetics of congenital hypothyroidism 381

mutation may contribute to thyroid dysfunction in that genetic cause for thyroid agenesis in humans. Subsequently,
population, though this is very unlikely in the homozygous two siblings were studied from an unrelated consanguineous
state. family, with a milder phenotype consisting of thyroid
There have also been reports of mildly raised TSH levels in agenesis, cleft palate, and spiky hair, but with absence of
individuals who are heterozygous for loss of function choanal atresia and bifid epiglottis (fig 2).44 They were
germline mutations in the TSHR.19 27 30–32 All have been homozygous for a different missense mutation (S57N) in the
reported to have normal sized eutopic thyroid glands on forkhead domain of TTF-2, and the S57N mutant protein
ultrasound scanning, except one who had thyroid hypopla- showed only partial loss of DNA binding and transcriptional
sia.32 The clinical phenotype of these individuals is not readily activity in vitro, possibly explaining the incomplete clinical
distinguishable from those with compensated TSH resistance phenotype.
who are homozygous or compound heterozygous for muta- TTF-1 is a homeobox transcription factor of the NK-2 gene
tions in the TSHR gene. Thus in families where non- family which is related to Drosophila NK-2/vnd (ventral
autoimmune subclinical hypothyroidism appears to be nervous system defective).45 The homeobox gene superfamily
inherited in an autosomal dominant manner, the possibility encodes transcription regulatory proteins that act at critical
of a heterozygous mutation in the TSHR should be points in development and ontogeny by sequence specific
considered. DNA binding, mediated by a structurally conserved homeo-
domain.46 TTF-1 has two independent transcriptional activa-
THYROID DYSGENESIS AND SYNDROMIC tion domains located at the amino terminal (N domain) and
CONGENITAL HYPOTHYROIDISM the carboxy terminal (C domain) regions with respect to the
Evidence to date suggests that the development of the DNA binding homeodomain.47 The human locus is found on
embryonic thyroid gland and its normal migration is chromosome 14q13, and the gene (also known as NKX-2.1)
dependent on the interplay between several transcription comprises three exons encoding a 42 kDa protein.48
factors. In the thyroid gland, the target genes of these Initial evidence of a role for TTF-1 (also known as thyroid
transcription factors include the thyroglobulin and thyroid specific enhancer binding protein, T/EBP) in the aetiology of
peroxidase (TPO) genes. Transcription factors that have been congenital hypothyroidism also came from a report of a
studied in human congenital hypothyroidism include TTF-1, mouse model in which the Titf-1 gene had been disrupted by
TTF-2, and the paired homeodomain factor Pax-8. In mice, homologous recombination.49 Heterozygous animals were
the expression of TTF-1, TTF-2, and Pax-8 begins at the onset initially described as having a normal euthyroid phenotype
of thyroid migration on day 9.5 of gestation and these factors but more recently were found to have reduced motor
continue to be expressed throughout embryonic develop- coordination skills when compared with wild type mice.50
ment.15 36 37 The onset of thyroid differentiation is heralded by Mice homozygous for the Titf-1 gene knockout were born
the expression of TSHR, TPO, and TG on day 14.5 of mouse dead, lacking lung parenchyma, with absent thyroid and
gestation. entire pituitary glands, and with extensive defects in the
TTF-2 is a transcription factor that is a member of the ventral forebrain.
forkhead/winged helix domain protein family, many of Not unexpectedly, TTF-1 is expressed in the lungs and
which are key regulators of embryonic pattern formation ventral forebrain, in addition to the thyroid gland.51 It is
and regional specification.36 It is a phosphoprotein that known to regulate the transcription of TG and TPO genes, the
consists of an N terminal region, a highly conserved forkhead TSHR gene in thyroid follicular cells,52 53 and the surfactant
domain, an a helical polyalanine tract, and unique C terminal protein B (SPB) gene in epithelial lung cells.54 A role for TTF-1
residues.38 Unlike some other transcription factors, such as in human respiratory development has been borne out by
HOX D13, polymorphism in the size of the polyalanine tract reports of heterozygous de novo TITF-1 deletions (14q13–21
in TTF-2 has been shown not to affect its transcriptional and 14q12–13.3, encompassing the TITF-1 locus) and a
function.38 The human gene is located in chromosome 9q22 mutation (at the 39 splice consensus of intron 2) inherited in
and consists of a single exon.39 Animal studies have an autosomal dominant manner being associated with
demonstrated the critical role of TTF-2 in thyroid embryonic compensated congenital hypothyroidism and unexplained
development. Heterozygous Titf-2 knockout mice are euthyr- respiratory distress in term babies in the neonatal period who
oid, with no visible phenotype, whereas homozygous null had normal bronchial morphology.55–57 The probands had
mice have cleft palate and thyroid dysgenesis, consisting of normal sized eutopic thyroid glands on radionuclide and
either thyroid agenesis or an ectopic sublingual gland, which ultrasound scanning, and asymmetrical 99mTc uptake was
is often lethal in the neonatal period.40 In mouse embryos, noted in one report.57 The other prominent recurrent features
Titf-2 is known to be expressed not only in the thyroid gland associated with either de novo or dominantly inherited
but also in the craniopharyngeal ectoderm involved in palate TITF-1 mutations (including one interstitial deletion in
formation and in Rathke’s pouch.36 More recently, its chromosome 14q) are neurological and include hypotonia,
expression has also been demonstrated in pharyngeal persistent ataxia and dysarthria, microcephaly, choreoathe-
endoderm derivatives, such as tongue, palate, epiglottis, tosis, and global developmental delay, suggesting a role for
and oesophagus, and in choanae and whisker follicle in the TTF-1 in human brain development.57–59 Major feeding
mouse and in human thyroid, hair follicle, and prepubertal difficulties were also noted. It is thought that the unfavour-
testis.41 42 able neurological outcome was most probably related to
Two Welsh male siblings with a constellation of defects TTF-1 deficiency in the thyroid and brain rather than to
similar to those in the homozygous knockout mice (con- inadequately corrected congenital hypothyroidism or inade-
genital hypothyroidism associated with thyroid agenesis and quate thyroxine replacement in an affected mother during
cleft palate)—with added features of spiky hair, bilateral pregnancy. Further data published recently suggest that
choanal atresia, and hypoplastic bifid epiglottis43—were TITF-1 mutations, leading to haploinsufficiency, are also
investigated for defects in human TITF-2 (also known as associated with isolated benign hereditary chorea, an
FKHL15 or FOXE1) and found to be homozygous for a autosomal dominant movement disorder.50
missense mutation (A65V) within its highly conserved Pax-8 is a transcription factor that is one of the nine
forkhead DNA binding domain.39 The mutant TTF-2 protein members of the mammalian paired homeodomain family
showed impaired DNA binding and loss of transcriptional which recognise DNA through the conserved paired domain
function. This report represented the first description of a and are homologous to the Drosophila segmentation genes.60

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
382 Park, Chatterjee

Figure 2 Patients with TITF-2 mutations, showing spiky hair, micrognathia, and hypertelorism (A), and cleft palate (B). (From: Castanet M, et al. A
novel loss-of-function mutation in TTF-2 is associated with congenital hypothyroidism, thyroid agenesis, and cleft palate. Hum Mol Genet
2002;11:20521–9; reproduced by permission of Oxford University Press.)

PAX genes perform key functions in mammalian embryonic non-syndromic or syndromic congenital hypothyroidism.
development, during which they show highly restricted Outside the thyroid gland, Pax-8 is also expressed in the
temporal and spatial expression patterns.61 Pax-8 has a kidney, where it is known to activate the Wilms’ tumour gene
DNA binding domain at the amino terminal end, a carboxy (WT1) promoter, and in the developing brain.70 Marked
terminal transcriptional activation domain, and a central phenotypic variability has been found within affected
homeodomain.62 The PAX-8 gene maps to human chromo- families, suggesting varied penetrance and expressivity of
some 2q12-q14 and consists of 11 exons.63 Studies have PAX-8 gene defects.
shown that Pax-8 plays a fundamental role not only in the All mutations to date have been located in the conserved
initiation of thyroid cell differentiation but also in the paired domain of PAX-8, and the mutant proteins have been
maintenance of the differentiated state, and that it is shown to have markedly reduced DNA binding with
essential for thyroid cell proliferation.64 Once again, mouse subsequent loss of transcriptional activation function. At
models have been instrumental in demonstrating the critical the structural level, these mutations are thought to disrupt
role of Pax-8 in thyroid organogenesis and thyroid cell the pronounced gain of a helical content following interac-
differentiation. Pax-8 null mice have hypoplastic thyroid tion of Pax-8 with DNA—that is, impairment of the
glands with absent median anlage derivatives (that is, unstructured to structured transition that occurs during
follicular cells), whereas lateral anlagen derivatives (para- DNA recognition (loss of ‘‘induced fit’’).68 Pax-8 activates
follicular calcitonin producing C cells) are present.65 transcription of TPO, TG, and NIS, and acts synergistically
The survival of homozygous mutant mice depends on with TTF-1 to activate the promoter of human TG gene.70–72
thyroxine replacement therapy, whereas heterozygous mice Dominant negative properties of the mutant allele or
do not display an obvious thyroid phenotype. In contrast, haploinsufficiency are not considered likely underlying
heterozygous human PAX-8 mutations have been described in mechanisms for pathogenicity of Pax-8 mutations. The exact
patients with congenital hypothyroidism of varying severity mechanism is still unclear but one possibility is that the
in six families: four familial cases where congenital human PAX-8 locus is imprinted, with selective expression of
hypothyroidism appears to be inherited in an autosomal the mutant allele leading to disease.
dominant manner and two sporadic cases.66–69 The thyroid The stimulatory G protein a subunit gene (GNAS1) is
glands in these patients are hypoplastic and sometimes located on chromosome 20q13 and contains over 13 exons
ectopic in location. In one family, the thyroid gland was of that encode Gsa, the a subunit of the heterotrimeric
normal size at birth but failed to develop normally stimulatory G protein which has intrinsic GTPase activity.73
postnatally, becoming hypoplastic.69 In this family, the G proteins mediate signal transduction across cell mem-
probands also showed decreased iodide trapping, with branes, coupling extracellular receptors—including those
a positive perchlorate discharge test in one, initially sug- binding TSH, TRH, parathyroid hormone (PTH), and luteinis-
gestive of dyhormonogenesis as a basis for congenital hypo- ing hormone—to intracellular effector proteins such as ion
thyroidism. channels and the adenylyl cyclase and phospholipase C
TPO is known to be particularly dependent on Pax-8 for second messenger systems.74 Heterozygous inactivating
efficient transcription, and therefore reduced TPO expression mutations in GNAS1 result in Albright hereditary osteo-
secondary to impaired Pax-8 function may be an explanation dystrophy. This is an autosomal dominant disorder char-
for the partial organification defect.64 Renal hemiagenesis acterised by recognisable dysmorphic features including short
was also reported in two affected cases, one being associated stature, shortened fourth and fifth metacarpals and meta-
with hypercalciuria.59 69 When present, renal agenesis was tarsals, obesity, subcutaneous ossification, intracranial calci-
ipsilateral to thyroid hemiagenesis.59 Thus congenital fication, and variable mental retardation. Presumably owing
hypothyroidism caused by PAX-8 mutations can occur as to imprinting mechanisms there is an apparent parent of

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
Genetics of congenital hypothyroidism 383

origin effect whereby maternal transmission usually leads to TPO, the enzyme responsible for iodide oxidation, organi-
a pseudohypoparathyroidism type Ia (PHPIa) phenotype, fication, and iodotyrosine coupling, is a 933 amino acid,
while mutations of paternal origin result in pseudopseudo- membrane bound, glycated, haem containing protein, located
hypoparathyroidism (PPHP).75 PHPIa consists of end organ on the apical membranes of the thyroid follicular cell.77 The
resistance to multiple hormones including PTH and TSH, human TPO gene is located on chromosome 2p25 and covers
leading to low serum calcium and raised levels of phosphate, approximately 150 kb of DNA.78 The most prevalent cause of
PTH, and TSH, whereas PPHP patients have Albright dyshormonogenesis is TPO deficiency.79 Defects in the TPO
hereditary osteodystrophy with a normal biochemical profile. gene have been reported to cause congenital hypothyroidism
The degree of TSH resistance tends to be mild, and overt by a total iodide organification defect.80 Increasing numbers
clinical hypothyroidism is not always present. The TSH of mutations—including maternal isodisomy for chromo-
response to TRH is exaggerated and the degree of TSH some 2p—have been reported, occurring throughout the 17
resistance may increase with age.76 exons of the thyroid peroxidase gene, each resulting in an
inactive TPO protein.80–91 There is one report of metastatic
DYSHORMONOGENESIS AND CONGENITAL thyroid carcinoma arising within a congenital goitre asso-
ciated with a mutation in the TPO gene.85
HYPOTHYROIDISM
Thyroglobulin is a homodimer with subunits of 330 000
The end product of a normally developed hypothalamic-
Da, which is synthesised exclusively in the thyroid gland. The
pituitary-thyroid axis is the production of thyroid hormones.
Iodide is actively transported and concentrated in the thyroid human gene is greater than 300 000 bp in size and is located
gland by the sodium iodide symporter present in the on chromosome 8q24.92 The coding sequence is divided into
basolateral membrane of thyroid follicular cells (fig 3). 42 exons, each of about 200 bp in size, except that exon 9 and
Subsequently it is oxidised by hydrogen peroxide and bound 10 contain 1101 p and 588 bp, respectively.77 Thyroglobulin
to tyrosine residues in thyroglobulin to form iodotyrosine defects arising from mutations in the gene are associated
(iodide organification). Some of these hormonally inert with moderate to severe congenital hypothyroidism, usually
iodotyrosine residues (monoiodotyrosine and diiodotyrosine) with low serum thyroglobulin concentrations.93 Affected
couple to form the hormonally active iodothyronines, T4 and individuals often have abnormal iodoproteins in their serum,
T3. Thyroid peroxidase (TPO) catalyses the oxidation, especially iodinated albumin, and they excrete iodopeptides
organification, and coupling reactions. Defects in any of of low molecular weight in the urine.77 There is ineffective
these steps lead to dyshormonogenesis which typically formation of T4 and T3 resulting from a coupling defect.
manifests as congenital hypothyroidism and goitre,77 and Owing to its size (coding sequence of 8244 bp), sequencing of
the responsible gene at each step has been cloned. Except in the TG gene has been difficult. However, increasing numbers
rare cases, all mutations in these genes appear to be inherited of mutations are being reported.94 95 Thyroglobulin is nor-
in an autosomal recessive fashion. mally exported through the secretory pathway into the lumen
of thyroid follicles to undergo iodination. Probands with
deficient thyroglobulin resulting from an altered TG coding
sequence have shown defective trafficking of thyroglobulin
TSH from the endoplasmic reticulum to Golgi, leading to an
Basolateral I– endoplasmic reticulum storage disease (ERSD).96 A particular
NIS T4, T3
membrane feature of some TG mutations involving cysteine residues in
TSHR
the protein is disruption of the three dimensional structure of
the molecule, causing it to be retained in the endoplasmic
reticulum as aggregates.96 The cog/cog mouse with a point
H2O2 I– mutation in the tg gene, in a region which is strictly
Thyroid follicular cell
conserved in the thyroglobulin proteins from all known
species, is an example of an ERSD and a model for severe
I congenital hypothyroidism associated with colloid deficient
Pendrin goitre and abnormal growth and central nervous system
I–
development.97 These mice have aberrant folding, dimerisa-
ThOX TPO tion, and export of thyroglobulin, leading to an abnormally
distended endoplasmic reticulum, comparable to that
Iodotyrosine
TG observed in thyroid biopsies from children with congenital
I– goitre from defective thyroglobulin synthesis.98 99
Apical Iodotyrosine NIS is an integral membrane protein of approximately 65
TPO kDa with 12 potential transmembrane domains,100 with both
membrane
T4, T3
carboxy and amino termini located inside the cell. Its
Lumen expression has not only been detected in normal and
neoplastic thyroid but also in salivary glands, gastric mucosa,
Figure 3 Schematic diagram of a follicular cell, illustrating the steps breast, colon, ovaries (lower species), placenta, skin, and
involved in thyroid hormone synthesis. TSH receptor (TSHR) bound to choroid plexus.101 The human gene (NIS) has been mapped to
TSH stimulates iodide transport into the thyroid gland by the sodium chromosome 19p, and the coding region contains 15 exons
iodide symporter (NIS). Subsequently, iodide is oxidised by hydrogen
encoding a protein of 643 amino acids.101 Before the cloning
peroxide, generated by the recently discovered NADPH oxidase system
(ThOX) and bound to tyrosine residues in thyroglobulin (TG) to form of NIS, a clinical diagnosis of hereditary iodide transport
iodotyrosine (iodide organification). Some of these hormonally inactive defect had been made for several decades on the basis of
iodotyrosine residues (monoiodotyrosine and diiodotyrosine) couple to goitrous hypothyroidism and absent thyroidal radioiodine
form the hormonally active iodothyronines, T4 and T3. Thyroid uptake.102 The first demonstration of a loss of function
peroxidase (TPO) catalyses the oxidation, organification, and coupling mutation in the NIS gene was reported in 1997, following
reactions. The exact function of pendrin, a chloride-iodide transporter, in
thyroid hormone synthesis is as yet unknown but it is thought to transport
which several mutations inherited in an autosomal recessive
iodide into the colloid from the thyrocyte. Defects in any of these steps manner have been described.103 The hypothyroidism is of
lead to dyshormonogenesis, which manifests clinically as congenital variable severity (ranging from fully compensated to severe)
hypothyroidism with goitre. and goitre is not always present. Individuals with a higher

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
384 Park, Chatterjee

dietary iodine intake are less likely to have severe hypothyr- prohormone as it is converted to the biologically more active
oidism than those with iodine deficient diets.104 This hormone triiodothyronine (T3) by 59-monodeiodination in all
condition is therefore preferably treated with iodine supple- tissues. In a negative feedback loop, T3 suppresses TSH
mentation rather than thyroid hormone replacement. People secretion. Thyroid hormone exerts its effects by acting on
who are heterozygous for a mutation are euthyroid.103 The nuclear receptors, necessitating transmembrane passage of
loss of function associated with some of these mutations the hormone. Several classes of membrane thyroid hormone
(Q267E and S515X) was accounted for by failure of transporters have been identified recently, including MCT8,
membrane targeting by the transporter.105 which was mapped to Xq13.2 and contains five exons.118
Pendred syndrome is an autosomal recessive disorder There are recent reports of mutations in this gene found in
characterised by sensorineural deafness and goitre that was male individuals in four families with abnormal thyroid
first described by Vaughn Pendred in 1896. The incidence of function tests consisting of low FT4 and raised levels of FT3
the disease is estimated to be 7.5 to 10 in 100 000, and it is and TSH, which were not found at the time of neonatal
thought to account for as many as 10% of cases of hereditary screening, but detected within the first two years of life.119
deafness, making it the most common cause of syndromic Stigmata of congenital hypothyroidism were not present at
deafness.106 Deafness is classically associated with a Mondini birth. In addition, neurological abnormalities from early
cochlear defect, consisting of a reduced number of turns in infancy were described, consisting of central hypotonia,
the cochlea, together with an enlarged vestibular aqueduct; peripheral hypertonia, dystonia, rotary nystagmus, dysconju-
these features are typically present at birth.107 108 Thyroid gate eye movements, feeding difficulties, vomiting, recurrent
disease usually presents as a multinodular or diffuse goitre of aspiration, irritability, and subsequent spastic quadriplegia
varying size in most affected individuals, and is typically not resulting in severe delay in motor and mental development
evident until the second decade of life. Despite the goitre, with absent speech reported in one proband. Brain MRI and
individuals are likely to be euthyroid and only rarely present electroencephalography were normal. Thyroid imaging has
with congenital hypothyroidism.109 TSH levels, however, are not been described. Female relatives harbouring mutations in
often in the upper end of the normal range, and hypothyr- MCT8 displayed milder thyroid phenotypes without the
oidism of variable severity may eventually develop.106 neurological features. Expression of this gene has been found
Intrafamilial phenotypic variation has also been noted.110 in all tissues examined, including brain, thyroid, pituitary,
Affected subjects usually have a positive perchlorate dis- and placenta.119 This is the first example of X linked
charge test, with more than 15%—but not complete—release congenital hypothyroidism resulting from a defect in target
of radiolabelled iodine following perchlorate administration, tissue rather than in the pituitary-thyroid biosynthetic
indicating a mild thyroid organification defect.106 A normal pathway.
discharge test, on the other hand, does not exclude the
diagnosis.110 The PDS gene is on chromosome 7q, contains 21 INVESTIGATION AND MANAGEMENT OF
exons, and is found to be expressed in the cochlea as well as
CONGENITAL HYPOTHYROIDISM
in the thyroid.111 It encodes pendrin, a 780 amino acid protein
In those cases of congenital hypothyroidism where an
(molecular weight 86 kDa) with 11 transmembrane domains
underlying genetic defect is suspected—on the basis of
which functions as a chloride-iodide transporter.112 In
family history, evidence of dyshormonogenesis, or the
contrast to the basolateral cell surface expression of NIS,
presence of other congenital anomalies—to suggest a
pendrin has been localised to the apical membrane of a
syndromic form of the disorder, further investigation can
subset of thyroid follicles.112 It has been hypothesised that
prove fruitful. While the outcome of such genetic analysis
pendrin transports iodide across the apical membrane of the
may not necessarily affect the management of the index
thyrocyte into the colloid space and that this process is
patient, the results can aid genetic counselling regarding
disrupted in Pendred syndrome, such that iodide is taken up
recurrence risk within the family and may suggest the best
normally by the thyrocyte but is not efficiently bound to
treatment option—for example, congenital hypothyroidism
thyroglobulin in the colloid.113
from NIS defects is more amenable to treatment with iodide
NADPH oxidases—encoded by two recently cloned genes,
supplementation than with thyroxine. We suggest below
THOX1 and THOX2, located at the apical membrane of
some investigations to aid in the search for an underlying
thyrocytes—are involved in H2O2 generation in the thyroid.114
genetic cause in a small number of cases with congenital
Both genes are highly expressed in thyrocytes.114 Thyroid
hypothyroidism. They are not intended to be a comprehen-
peroxidase (TPO) has no biological activity in the absence of
sive list of recommended investigations for physicians
H2O2, which is likely to be the limiting factor for thyroglo-
managing routinely diagnosed cases.
bulin iodination when iodide supply is normal.115 The two
As congenital hypothyroidism is largely a sporadic dis-
human THOX genes are arranged in a head to head
order, a family history is not present in most cases. However,
configuration and are separated by a 16 kb long region.116
a detailed history should be taken, including that of parental
They span 75 kb and are composed of 35 and 34 exons,
consanguinity, other affected family members, and a history
respectively. How THOX participates in Ca2+/NADPH depen-
of any extrathyroidal congenital anomalies (for example,
dent H2O2 generation in thyroid tissue is still unknown.
cleft palate, renal anomalies, neonatal respiratory distress,
Defects in the human THOX2 have recently been reported,
and movement disorders). A neonate born with severe
with heterozygous truncation mutations being associated
congenital hypothyroidism may have the associated features
with mild transient congenital hypothyroidism and a partial
of prolonged jaundice, macroglossia, hoarse cry, and umbi-
iodide organification defect, whereas homozygosity for such
lical hernia on examination. The neck should be examined
defects was associated with severe congenital hypothyroid-
for a goitre and its presence would suggest dyshormonogen-
ism and a complete iodide organification defect.117 Goitre was
esis as the basis of congenital hypothyroidism. Rarely, the
not detected in these patients.
goitre can be detected in utero by ultrasound scanning, and
such cases can be treated antenatally by intra-amniotic
IODOTHYRONINE TRANSPORTER DEFECTS AND injections of thyroxine or triiodothyronine, which can
SYNDROMIC CONGENITAL HYPOTHYROIDISM successfully shrink the gland.120 If a significantly enlarged
TSH secreted by thyrotrophs in the anterior pituitary gland goitre escapes antenatal detection, there could be acute
stimulates the gland to synthesise and secrete thyroid problems postnatally if the upper airway is compromised, and
hormones, principally thyroxine (T4). T4 is essentially a the expert opinion of a paediatric ENT specialist is vital in

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
Genetics of congenital hypothyroidism 385

Transient
? Material anti-TSH receptor antibody
CH
? THOX2
Permanent

History, examination
TFTs, serum TG
Thyroid scintigraphy
Ultrasound scan
Negative perchlorate discharge test
Perchlorate discharge test

Dyshormonogenetic CH Non-dyshormonogenetic CH
Positive perchlorate discharge test
Gland but
↓Serum TG poor tracer
TIOD uptake
Extra-thyroid anomalies
Hearing
loss

? TG ? PDS ? TPO ? NIS Absent


? THOX2
Present
? TSHR
? PAX-8

Cleft palate/spiky hair → ? TITF-2


Renal anomaly → ? PAX-8
Neonatal respiratory distress/chorea/ataxia → ? TITF-1
AHO → ? GNAS1
Other anomalies → ? novel candidate genes

Figure 4 A proposed algorithm for investigating the genetic basis of congenital hypothyroidism. AHO, Albright hereditary osteodystrophy; CH,
congenital hypothyroidism; GNAS, stimulatory G protein a subunit gene; NIS, sodium-iodide symporter gene; PAX-8, human Pax-8 gene; PDS,
Pendred syndrome gene; TFTs, thyroid function tests; TG, thyroglobulin gene; THOX2, thyroid oxidase 2; TIOD, total iodide organification defect;
TITF-1, human TTF-1 gene; TITF-2, human TTF-2 gene; TPO, thyroid peroxidase gene; TSHR, TSH receptor gene; USS, ultrasound scan.

such cases. The presence of any other congenital malforma- measurement of serum thyroglobulin is also useful as all
tions should also be sought on examination. Delayed patients with organification disorders, and cases with severe
maturation of bone on x ray is of limited value in infancy TSHR defects, have raised levels when their circulating TSH is
as the ossification centres are composed mainly of cartilage; it increased, except in cases with thyroglobulin synthesis defects
is of greater value after two years in severe forms of where the thyroglobulin level is usually low. Thyroglobulin
congenital hypothyroidism. The initial crucial steps in measurements can provide useful additional information about
investigating congenital hypothyroidism are to establish the the thyroid gland—for example, if serum thyroglobulin is
severity of the hormone defect, to investigate for the measurable where there is apparent athyreosis on thyroid
possibility of dyshormonogenesis, and determine the mor- imaging, this suggests that radiologically undetectable hypo-
phology and position of the thyroid gland (fig 4). plastic thyroid tissue is likely to be present.32
It is also worth checking thyroid function (circulating TSH,
Biochemical investigations FT4, and FT3) and thyroglobulin levels not only in affected
The first of these steps is established by obtaining the original family members but also in all first degree relatives of
diagnostic thyroid function tests from the neonatal blood probands, as variable penetrance associated with autosomal
spot screening card and from the first venous serum inheritance might lead to the identification of a mildly
measurements, before the onset of L-thyroxine treatment. affected relative not detected previously. Furthermore, with
In up to 10% of cases, the congenital hypothyroidism is TSHR mutations, heterozygotes may also display a mild
transient and self limiting, lasting up to a few months.121 phenotype. Underlying autoimmune thyroid disease, causing
Some of these cases can be accounted for by the presence of raised TSH with low or normal thyroid hormones, can be
anti-TSHR antibodies in the mother, with transplacental excluded by the absence of thyroid autoantibodies. A useful
passage to the fetus. When detected, the function of these test to determine whether the thyroid gland is resistant to
TSHR autoantibodies should be measured, as there are TSH involves the administration of TRH, with measurement
reports of hyperthyroid mothers with Graves’ disease giving of both the pituitary TSH response and the subsequent rise in
birth to hypothyroid infants.122 circulating T3 following this increase in TSH.
In other infants with transient congenital hypothyroidism Defective iodide trapping by the thyroid gland caused by
and an abnormal perchlorate discharge test suggestive of a NIS mutations can be tested for by measuring the saliva to
defect in organification within the thyroid, the possibility of plasma (S/P) ratio of iodide one hour after oral administra-
heterozygous mutations in THOX2 should be considered. The tion of a small dose of radiolabelled iodide (between 25 and

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
386 Park, Chatterjee

50 mCi of either 125I or 131I labelled NaI). The normal S/P ratio its involvement in recessive disease, and possibly also
is 25 to 140.123 Thyroxine treatment need not be stopped or heterozygous mutations in this gene in congenital or even
reduced before this test, as it does not affect salivary gland acquired non-autoimmune hypothyroidism. Downstream of
iodide trapping. TSHR, defects in Gsa result in TSH resistance in Albright’s
hereditary osteodystrophy. Defects in the transcription
Radiological investigations factors TITF-2 (cleft palate, spiky hair), TITF-1 (neonatal
Thyroid scintigraphy, using either technetium (Tc) or radio- respiratory distress, involuntary movement), and Pax-8 (renal
active iodine, is usually the first line imaging in many centres hemiagenesis) provide the basis for multisystem involvement
and provides information about the size and location of the in syndromic forms of congenital hypothyroidism. Finally,
gland, but it can be misleading. For example, it can indicate there is early evidence emerging for a third group of
no uptake of isotope, suggesting apparent athyreosis, congenital hypothyroid conditions associated with defects
although a thyroid gland is present. Ultrasound scanning of in target tissue iodothyronine transporters, with devastating
the neck by an experienced radiologist can provide further neurological features.
valuable information, with assessment of whether thyroid Novel and as yet unidentified target genes regulated by
gland size is appropriate for age and sex, and its location these transcription factors may also prove to be important
along the embryological line of development between the candidate genes in other forms of congenital hypothyroidism.
base of the tongue and thorax. Ultrasound scanning in A role for the transcription factor Hoxa3 (human locus
conjunction with isotope scanning can provide further useful 7p15.1), a member of the Hox family of homeobox genes
information. For example, if there is discordance between involved in regulating the development of pharyngeal
thyroid isotope scanning and ultrasound scanning, with the glandular organs, has hitherto only been shown in murine
former showing no tracer uptake yet the latter showing the embryonic thyroid development, with null mice having
presence of a normal or even an enlarged thyroid gland, there thyroid hypoplasia resulting from defects in both follicular
is strong evidence for the involvement of NIS causing and parafollicular cell development.129 Interestingly, these
congenital hypothyroidism. Based on history and initial mice also have other abnormalities, including thymic and
investigations, further imaging can be undertaken, such as parathyroid aplasia, anomalies of the heart and great vessels,
a renal ultrasound scan in a family with autosomal dominant and malformations of the throat and jaw, which are
congenital hypothyroidism associated with thyroid gland remarkably similar to those observed in 22q11 deletion
hypoplasia (?Pax-8 defect), or MRI of the inner ear in cases of (velocardiofacial) syndrome in humans.130 It is also known
congenital hypothyroidism with goitre and hearing loss. The that the amount of thyroid tissue in these patients is variably
proportion of radioiodine discharged from the thyroid reduced, as in the Hoxa3 mutant mice,131 and it has therefore
following the administration of potassium perchlorate is an been suggested that patients with 22q11 deletion syndrome
index of the severity of organification defects. Discharge of may be at increased risk of thyroid dysfunction. Hoxa5(2/2)
about 20–45% indicates a partial and mild defect, but values mutant mice have been reported to have hypothyroidism
greater than 60% and 90% are typical of severe and complete associated with transient growth retardation, delayed eye
defects, respectively.80 opening, and ear elevation.132 Thyroid gland development
begins normally, but follicle formation and thyroglobulin
Treatment processing are abnormal in late gestation. The expression of
Once the diagnosis of congenital hypothyroidism is made on several molecular markers essential for thyroid gland forma-
neonatal screening, appropriate thyroxine replacement ther- tion and function—namely TTF-1, Pax8, and TTF-2—is
apy is initiated by a paediatrician. Studies have shown that affected in the developing thyroid gland, with the conse-
several variables influence eventual IQ in children with quence of altered expression of thyroid effector genes,
congenital hypothyroidism. These include the severity of including the thyroglobulin and TPO genes. Murine Nkx-
congenital hypothyroidism (as determined by thyroid func- 2.5, a member of the homeobox gene superfamily that is
tion tests and delayed skeletal maturation at birth), the related to the TITF1 (NKX-2.1), is expressed early during
dose of thyroxine treatment, the timing of treatment onset, embryogenesis of both thyroid and myocardium,133 and
and serum free thyroxine concentrations during the first accordingly represents a strong candidate in those 4% of
year.124–127 Despite the establishment of neonatal screening cases of congenital hypothyroidism that are associated with
programmes, clinicians and families should be aware of cardiac defects.134
reports suggesting that about 10% of early treated infants With regard to identification of additional novel candidate
with severe hypothyroidism are likely to require special genes, future directions include genome-wide linkage ana-
education.128 Therefore clinically significant intellectual lyses in large families with multiple probands or parental
impairment should be actively screened for and treated consanguinity, identification of congenital hypothyroidism
when detected in these children. phenotypes in transgenic or randomly mutagenised mouse
models, and linkage analysis and positional cloning in well
characterised contiguous gene syndromes with congenital
CONCLUSIONS hypothyroidism as a prominent feature (for example, William
Congenital hypothyroidism represents a common neonatal syndrome (7q11 deletion) where congenital hypothyroidism
problem that is easily detected and treated, and hence the occurs in 25% of affected individuals). Known candidate
success of the nationwide neonatal screening programmes. genes will have to be carefully ruled out in investigating for
While the great majority of cases are considered sporadic, this genetically heterogeneous condition where phenocopies
there have been recent advances in elucidating some of the pose difficulties—for example, in the absence of a family
molecular mechanisms behind certain forms of this common history it may be difficult to distinguish between TSHR
congenital metabolic disorder. Organification defects leading mutations and Pax-8 defects affecting only the thyroid.
to goitrous congenital hypothyroidism are now well known to
have an autosomal recessive genetic basis. More recently,
ACKNOWLEDGEMENTS
there is growing evidence for the role of germline gene SMP is a recipient of a Wellcome Trust research training fellowship
defects causing congenital hypothyroidism associated with for medical and dental graduates and the Raymond and Beverly
thyroid dysgenesis. These include a role for the TSHR gene in Sackler studentship from the University of Cambridge School of
non-syndromic congenital hypothyroidism, with evidence for Clinical Medicine.

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
Genetics of congenital hypothyroidism 387

.....................
glucocorticoid deficiency is due to a mutation producing a truncated
Authors’ affiliations thyrotropin receptor. Thyroid 1999;9:887–94.
S M Park, Department of Clinical Genetics, Addenbrooke’s Hospital, 26 Tonacchera M, Agretti P, Pinchera A, Rosellini V, Perri, A, Collecchi P, Vitti P,
Cambridge, UK Chiovato L. Congenital hypothyroidism with impaired thyroid response to
V K K Chatterjee, Department of Medicine, University of Cambridge, thyrotropin (TSH) and absent circulating thyroglobulin: evidence for a new
Addenbrooke’s Hospital inactivating mutation of the TSH receptor gene. J Clin Endocrinol Metab
2000;85:1001–8.
Competing interests: none declared 27 Russo D, Betterle C, Arturi F, Chiefari E, Girelli ME, Filetti S. A novel mutation
in the thyrotropin (TSH) receptor gene causing loss of TSH binding but
constitutive receptor activation in a family with resistance to TSH. J Clin
REFERENCES Endocrinol Metab 2000;85:4238–42.
1 Toublanc J. Comparison of epidemiological data on congenital 28 Bretones P, Duprez L, Parma J, David M, Vassart G, Rodien P. A familial
hypothyroidism in Europe with those of other parts of the world. Horm Res case of congenital hypothyroidism caused by a homozygous mutation of the
(Basel) 1992;38:230–5. thyrotropin receptor gene. Thyroid 2001;11:997–8.
2 Grant DB, Smith I, Fuggle PW, Tokar S, Chapple J. Congenital 29 Nagashima T, Murakami M, Onigata K, Morimura T, Nagashima K, Mori M,
hypothyroidism detected by neonatal screening: relationship between Morikawa A. Novel inactivating missense mutations in the thyrotropin
biochemical severity and early clinical features. Arch Dis Child receptor gene in Japanese children with resistance to thyrotropin. Thyroid
1992;67:87–90. 2001;11:551–9.
3 Medeiros-Neto G, Stanbury JB, eds. Inherited disorders of the thyroid 30 Tonacchera M, Agretti P, De Marco G, Perri A, Pinchera A, Vitti P,
system. Boca Raton: CRC Press, 1994:1–221. Chiovato L. Thyroid resistance to TSH complicated by autoimmune thyroiditis.
4 Pintar JE. Normal development of the hypothalamic-pituitary-thyroid axis. In: J Clin Endocrinol Metab 2001;86:4543–6.
Braverman LE, Utiger RD, eds. Werner and Ingbar’s the thyroid, 7th ed. 31 Alberti L, Proverbio MC, Costagliola S, Romoli R, Boldrighini B, Vigone MC,
Philadelphia: Lippincott-Raven, 1996:6–18. Weber G, Chiumello G, Beck-Peccoz P, Persani L. Germline mutations of TSH
5 Williams E, Toyn C, Harach H. The ultimobranchial gland and congenital receptor gene as cause of nonautoimmune subclinical hypothyroidism. J Clin
thyroid abnormalities in man. J Pathol 1989;159:135–41. Endocrinol Metab 2002;87:2549–55.
6 Cao XY, Jiang XM, Dou ZH, Rakeman MA, Zhang ML, O’Donnell K, Ma T, 32 Park SM, Clifton-Bligh RJ, Betts P, Chatterjee VK. Congenital hypothyroidism
Amette K, DeLong N, DeLong GR. Timing of vulnerability of the brain to and apparent athyreosis with compound heterozygosity or compensated
iodine deficiency in endemic cretinism. N Engl J Med 1994;331:1739–44. hypothyroidism with probable hemizygosity for inactivating mutations of the
7 Klein RZ, Mitchell ML. Hypothyroidism in infants and children. In: TSH receptor. Clin Endocrinol (Oxf) 2004;60:220–7.
Braverman LE, Utiger RD, eds. Werner and Ingbar’s the thyroid, 7th ed. 33 Parma J, Duprez L, Van Sande J, Cochaux P, Gervy C, Mockel J, Dumont J,
Philadelphia: Lippincott-Raven, 1996:984–1008. Vassart G. Somatic mutation in the thyrotropin receptor gene cause
8 Castanet M, Polak M, Bonaiti-Pellie C, Lyonnet S, Czernickow P, Leger J. hyperfunctioning thyroid adenomas. Nature 1993;365:649–51.
Nineteen years of national screening for congenital hypothyroidism: familial 34 Duprez L, Parma J, Van Sande J, Allgeier A, Leclere J, Schvartz C, Delisle MJ,
cases with thyroid dysgenesis suggest the involvement of genetic factors. J Clin Decoulx M, Orgiazzi J, Dumont J, et al. Germline mutations in the
Endocrinol Metab 2001;86:2009–14. thyrotropin receptor gene cause non-autoimmune autosomal dominant
9 Grant DB, Smith I. Survey of neonatal screening for primary hypothyroidism hyperthyroidism. Nature Genet 1994;7:396–401.
in England, Wales, and Northern Ireland 1982–4. BJM 1988;296:1355–8. 35 Jordan N, Williams N, Gregory JW, Evans C, Owen M, Ludgate M. The
10 Law WY, Bradley DM, Lazarus JH, John R, Gregory JW. Congenital W546X mutation of the thyrotropin receptor gene: potential major
hypothyroidism in Wales (1982–1993): demographic features, clinical contributor to thyroid dysfunction in a Caucasian population. J Clin
presentation and effects on early neurodevelopment. Clin Endocrinol (Oxf) Endocrinol Metab 2003;88:1002–5.
1988;48:201–7. 36 Zannini M, Avantaggiato V, Biffali E, Arnone MI, Sato K, Pischetola M,
11 Strader CD, Fong TM, Tota MR, Underwood D. Structure and function of G Taylor BA, Phillips SJ, Simeone A, Di Lauro R. TTF-2, a new forkhead protein,
protein-coupled receptors. Annu Rev Biochem 1994;63:101–32. shows a temporal expression in the developing thyroid which is consistent
12 Vassart G, Dumont JE. The thyrotropin receptor and the regulation of with a role in controlling the onset of differentiation. EMBO J
thyrocyte function and growth. Endocrinol Rev 1992;13:596–611. 1997;16:3185–97.
13 Libert F, Passage E, Lefort A, Vassar G, Mattei MG. Localisation of human 37 Zannini M, Avantaggiato V, Biffali E, Arnone MI, Sato K, Pischetola M,
thyrotropin receptor gene to chromosome region 14q31 by in situ Taylor BA, Phillips SJ, Simeone A, Di Lauro R. TTF-2, a new forkhead protein,
hybridization. Cytogenet Cell Genet 1990;54:82–3. shows a temporal expression in the developing thyroid which is consistent
14 Gross B, Misrahi M, Sar S, Milgrom E. Composite structure of the human with a role in controlling the onset of differentiation [erratum]. EMBO J
thyrotropin receptor gene. Biochem Biophys Commun 1991;177:679–87. 2001;20:2108.
15 Damante G, Di Lauro R. Thyroid-specific gene expression. Biochem Biophys 38 Hishinuma A, Ohyama Y, Kuribayashi T, Nagakubo N, Namatame T,
Acta 1994;1218:255–66. Shibayama K, Arisaka O, Matsuura N, Ieiri T. Polymorphism of the
16 Stein SA, Oates EL, Hall CR, Grumbles RM, Fernandez LM, Taylor NA, polyalanine tract of thyroid transcription factor-2 gene in patients with
Puett D, Jin S. Identification of a point mutation in the thyrotropin receptor of thyroid dysgenesis. Eur J Endocrinol 2001;145:385–9.
the hyt/hyt hypothyroid mouse. Mol Endocrinol 1994;8:129–38. 39 Clifton-Bligh RJ, Wentworth JM, Heinz P, Crisp M, John R, Lazarus JH,
17 Postiglione MP, Parlato R, Rodiguez-Mallon A, Rosica A, Mithbaokar P, Ludgate M, Chatterjee VKK. Mutation of the gene encoding human TTF-2
Maresca M, Marians RC, Davies TF, Zannini MS, De Felice M, Di Lauro R. associated with thyroid agenesis, cleft palate and choanal atresia. Nat Genet
Role of the thyroid-stimulating hormone receptor signaling in development 1998;19:399–401.
and differentiation of the thyroid gland. Proc Natl Acad Sci USA 40 De Felice M, Ovitt C, Biffali E, Rodriguez-Mallon A, Arra C, Anastassiadis K,
2002;99:15462–7. Macchia PE, Mattei M-G, Mariano A, Scholer H, Macchia V, Di Lauro R. A
18 Evans C, Jordan NJ, Owens G, Bradley D, Ludgate M, John R. Potent mouse model for hereditary thyroid dysgenesis and cleft palate. Nat Genet
thyrotrophin receptor-blocking antibodies: a cause of transient congenital 1998;19:395–8.
hypothyroidism and delayed thyroid development. Eur J Endocrinol 41 Dathan N, Parlato R, Rosica A, De Felice M, Di Lauro R. Distribution of the
2004;150:265–8. titf2/foxe1 gene product is consistent with an important role in the
19 Sunthornthepvarakul T, Gootschalk ME, Hayashi Y, Refetoff S. Resistance to development of foregut endoderm, palate, and hair. Dev Dyn
thyrotropin caused by mutations in the thyrotropin-receptor gene. 2002;224:450–6.
N Engl J Med 1995;332:155–60. 42 Sequeira M, Al-Khafaji F, Park S, Lewis MD, Wheeler MH, Chatterjee VK,
20 De Roux N, Misrahi M, Brauner R, Houang M, Carel JC, Granier M, Le Jasani B, Ludgate M. Production and application of polyclonal antibody to
Bouc Y, Ghinea N, Boumeddienne A, Toublanc JE, Milgrom E. Four families human thyroid transcription factor 2 reveals thyroid transcription factor 2
with loss of function mutations of the thyrotropin receptor J Clin Endocrinol protein expression in adult thyroid and hair follicles and prepubertal testis.
Metabol 1996;81:4229–35. Thyroid 2003;13:927–32.
21 Clifton-Bligh RJ, Gregory JW, Ludgate M, John R, Persani L, Asteria C, Beck- 43 Bamforth JS, Hughes IA, Lazarus JH, Weaver CM, Harper PS. Congenital
Peccoz P, Chatterjee VKK. Two novel mutations in the thyrotropin (TSH) hypothyroidism, spiky hair, and cleft palate. J Med Genet 1989;26:49–60.
receptor gene in a child with resistance to TSH. J Clin Endocrinol Metabol 44 Castanet M, Park SM, Smith A, Bost M, Leger J, Lyonnet S, Pelet A,
1997;82:1094–100. Czernichow P, Charrerjee K, Polak M. A novel loss-of-function mutation in
22 Biebermann H, Schoneberg T, Krude H, Schuktz G, Gudermann T, TTF-2 is associated with congenital hypothyroidism, thyroid agenesis, and
Gruters A. Mutations in the human thyrotropin receptor gene causing thyroid cleft palate. Hum Mol Genet 2002;11:2051–9.
hypoplasia and persistent congenital hypothyroidism. J Clin Endocrinol 45 Harvey RP. NK-2 homeobox genes and heart development. Dev Biol
Metabol 1997;82:3471–80. 1996;187:203–16.
23 Abramowicz MJ, Duprez L, Parma J, Vassart G, Heinrich C. Familial 46 Scott MP, Tamkun JW, Hatzell GW. The structure and function of the
congenital hypothyroidism due to inactivating mutation of the thyrotropin homeodomain. Biochim Biophys Acta 1989;989:25–48.
receptor causing profound hypoplasia of the thyroid gland. J Clin Invest 47 De Felice M, Damante G, Zannini M, Francis-Lang H, Di Lauro R. Redundant
1997;99:3018–24. domains contribute to the transcriptional activity of thyroid transcription
24 Gagne N, Parma J, Deal C, Vassart G, Van Vliet G. Apparent congenital factor 1. J Biol Chem 1995;270:26649–56.
athyreosis contrasting with normal plasma thyroglobulin levels and 48 Guazzi S, Price M, De Felice M, Damante G, Mattei MG, Di Lauro R. Thyroid
associated with inactivating mutations in the thyrotropin receptor gene: are nuclear factor 1 (TTF-1) contains a homeodomain and displays a novel DNA
athyreosis and ectopic thyroid distinct entities? J Clin Endocrinol Metabol binding specificity. EMBO J 1990;9:3631–9.
1998;83:1771–5. 49 Kimura S, Hara Y, Pineau T, Fernandez-Salguero P, Fox CH, Ward JM,
25 Tiosano D, Pannain S, Vassart G, Parma J, Gershoni-Baruch R, Mandel H, Gonzalez FJ. The t/epb null mouse: thyroid-specific enhancer-binding
Lotan, R, Zaharan Y, Pery M, Weiss R, Refetoff S, Hochberg, Z. The protein is essential for the organogenesis of the thyroid, lung, ventral
hypothyroidism in an inbred kindred with congenital thyroid hormone and forebrain and pituitary. Genes Dev 1996;10:60–9.

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
388 Park, Chatterjee

50 Breedveld GJ, van Dongen JWF, Danesino C, Guala A, Percy AK, Dure LS, 78 Endo Y, Onogi S, Umeki K, Yamamoto I, Kotani T, Ohtaki S, Fujita T.
Harper P, Lazarou LP, et al. Mutations in TITF-1 are associated with benign Regional localization of the gene for thyroid peroxidase to human
hereditary chorea. Hum Mol Genet 2002;11:971–9. chromosome 2p25 and mouse chromosome 12C. Genomics
51 Lazzaro D, Price M, de Felice M, Di Lauro R. The transcription factor TTF-1 is 1995;25:760–1.
expressed at the onset of thyroid and lung morphogenesis and in restricted 79 Mangkalbruks A, Correa Billerbeck A-E, Wajchenberg B, Knobel M, Cox NJ,
regions of the foetal brain. Development 1991;113:1093–104. DeGroot LJ, Medeiros-Neto G. Genetic linkage studies of thyroid peroxidase
52 Civitreale D, Lonigro R, Sinclair AJ, Di Lauro R. A thyroid-specific nuclear (TPO) gene in families with TPO deficiency. J Clin Endocrinol Metab
protein essential for tissue-specific expression of the thyroglobulin promoter. 1991;72:471–6.
EMBO J 1989;8:2537–42. 80 Bikker H, Vulsma T, Baas F, de Vijlder JJM. Identification of five novel
53 Francis-Lang H, Price M, Polycarpou-Schwartz M, Di Lauro R. Cell-type- inactivating mutations in the human thyroid peroxidase gene by denaturing
specific expression of the rat thyroperoxidase promoter indicates common gradient gel electrophoresis. Hum Mutat 1995;6:9–16.
mechanisms for thyroid-specific gene expression. Mol Cell Biol 81 Abramowicz MJ, Targovnik HM, Varela V, Cochaux P, Krawiec L,
1992;12:576–88. Pisarev MA, Propato FV, Juvenal G, Chester HA, Vassart G. Identification of
54 Bruno MD, Bohinski RJ, Huelsman KM, Whitsett JA, Korfhagen TR. Lung cell- a mutation in the coding sequence of the human thyroid peroxidase gene
specific expression of the murine surfactant protein A (SP-A) gene is causing congenital goiter. J Clin Invest 1992;90:1200–4.
mediated by interactions between the SP-A promoter and thyroid 82 Bikker H, den Hartog MT, Baas F, Gons MH, Vulsma T, de Vijlder JJ. A 20-
transcription factor-1. J Biol Chem 1995;270:6531–6. basepair duplication in the human thyroid peroxidase gene results in a total
55 Devriendt K, Vanhole C, Matthijs G, de Zegher F. Deletion of the thyroid iodide organification defect and congenital hypothyroidism. J Clin
transcription factor-1 gene in an infant with neonatal thyroid dysfunction and Endocrinol Metab 1994;79:248–52.
respiratory failure. N Engl J Med 1998;338:1317–18. 83 Bikker H, Waelkens JJJ, Bravenboer B, de Vijlder JJJ. Congenital
56 Iwatani N, Mabe H, Devriendt K, Kodama M, Miike T. Deletion of NKX2.1 hypothyroidism caused by a premature termination signal in exon 10 of the
gene encoding thyroid transcription factor-1 in two siblings with human thyroid peroxidase gene. J Clin Endocrinol Metabol
hypothyroidism and respiratory failure. J Pediatr 2000;137:272–6. 1996;81:2076–9.
57 Doyle DA, Gonzalez I, Thomas B, Scavina M. Autosomal dominant 84 de Vijlder JJM, Bikker H, Ris-Stalpers C, Vulsma T. Structure, function, and
transmission of congenital hypothyroidism, neonatal respiratory distress, and relevance of thyroid peroxidase in inherited diseases of the thyroid. Curr
ataxia caused by a mutation of NKX2–1. J Pediatr 2004;145:190–3. Opin Endocrinol Diabetes 1997;4:328–32.
58 Pohlenz J, Dumitrescu A, Zundel D, Martine U, Schonberger W, Koo E, 85 Medeiros-Neto G, Gil-da-Costa MJ, Santos CS, Medina AM, Silva JCE,
Weiss RE, Cohen RN, Kimura S, Refetoff S. Partial deficiency of thyroid Tsou RM, Sobrinho-Simoes M. Metastatic thyroid carcinoma arising from
transcription factor 1 produces predominantly neurological defects in congenital goiter due to mutation in the thyroperoxidase gene. J Clin
humans and mice. J Clin Invest 2002;109:469–73. Endocrinol Metab 1998;83:4162–6.
59 Krude H, Schutz, Biebermann H, von Moers A, Schnabel D, Neitzel H, 86 Kotani T, Umeki K, Yamaoto I, Maesaka H, Tachibana K, Ohtaki S. A novel
Tonnies H, Weise D, Lafferty A, Schwarz S, DeFelice M, von Deimling A, van mutation in the human thyroid peroxidase gene resulting in a total iodide
Landeghem F, DiLauro R, Gruters A. Choreoathetosis, hypothyroidism, and organification defect. J Endocrinol 1999;160:267–73.
pulmonary alterations due to human NKX2-1 haploinsufficiency. J Clin Invest 87 Kotani T, Umeki K, Yamamoto I, Ohtaki S, Adachi M, Tachibana K. Iodide
2002;109:475–80. organification defects resulting from cosegregation of mutated and null
60 Stuart ET, Gruss P. PAX genes: what’s new in developmental biology and thyroid peroxidase alleles. Mol Cell Endocrinol 2001;182:61–8.
cancer? Hum Mol Genet 1995;4:1717–20. 88 Pannain S, Weiss RE, Kackson CE, Dian D, Beck JC, Sheffield VC, Cox N,
61 Mansouri A, Hallonet M, Gruss P. Pax genes and their role cell differentiation Refetoff S. Two different mutations in the thyroid peroxidase gene of a large
and development. Curr Opin Cell Biol 1996;8:851–7. inbred Amish kindred: power and limits of homozygosity mapping. J Clin
62 Poleev A, Fickenscher H, Mundlos S, Winterpacht A, Zabel B, Fidler A, Endocrinol Metab 1999;84:1061–71.
Gruss P, Plachov D. PAX-8, a human paired box gene: isolation and 89 Santos CL, Rego KG, Nascimento AC, Tambascia M, de Vijlder JJ, Medeiros-
expression in developing thyroid, kidney, and Wilm’s tumours. Development Neto G. A novel mutation in the TPO gene in goitrous hypothyroid patients
(Camb) 1992;116:611–23. with iodide organification defect. Clin Endocrinol (Oxf) 1999;51:165–72.
63 Poleev A, Wendler F, Gickenscher H, Zannini S, Yaginuma K, Abbott C, 90 Bakker B, Bikker H, Vulsma T, de Randamie JSE, Wiedijk BM, de Vijlder JJM.
Plachov D. Distinct functional properties of three human paired-box-proteins, Two decades of screening for congenital hypothyroidism in the Netherlands:
PAX8, isoforms generated by alternative splicing in thyroid, kidney and TPO gene mutations in total iodide organification defects (an update). J Clin
Wilms’ tumors. Eur J Biochem 1995;228:899–911. Endocrinol Metab 2000;85:3708–12.
64 di Magliano MP, Di Lauro R, Zannini M. Pax8 has a key role in thyroid cell 91 Ambrugger P, Stoeva I, Biebermann H, Torresani T, Leitner C, Gruter A.
differentiation. Proc Natl Acad Sci USA 2000;97:13144–9. Novel mutations of the thyroid peroxidase gene in patients with permanent
65 Mansouri A, Chowdhury K, Gruss P. Follicular cells of the thyroid gland congenital hypothyroidism. Eur J Endocrinol 2001;145:19–24.
require Pax8 gene function. Nat Genet 1998;19:87–90. 92 Berge-Lefranc JL, Cartonzon G, Mattei MG, Passage E, Malezet-
66 Macchia PA, Lapi P, Krude H, Pirro MT, Missero C, Chiovato L, Souabni A, Desmoulins C, Lissitzky S. Localisation of the thyroglobulin gene by in situ
Baserga M, Tassi V, Pinchera A, Fenzi G, Gruters A, Busslinger M, Di hybridization to human chromosomes. Hum Genet 1985;69:28–31.
Lauro R. PAX8 mutations associated with congenital hypothyroidism caused 93 DeGroot LJ. Congenital defects in thyroid hormone formation and action. In:
by thyroid dysgenesis. Nat Genet 1998;19:83–6. DeGroot LJ, ed. Endocrinology, 3rd ed. Philadelphia: WB Saunders,
67 Vilain C, Rydlewski C, Dupez L, Heinrichs C, Abramowicz, Malvaux P, 1995:871.
Renneboog B, Parma J, Costagliola S, Vassart G. Autosomal dominant 94 Targovnik HM, Frechtel GD, Mendive FM, Vono J, Cochaux P, Vassart G,
transmission of congenital thyroid hypoplasia due to loss-of-function Medeiros-Neto G. Evidence for the segregation of three different mutated
mutation of PAX8. J Clin Endocrinol Metab 2001;86:234–8. alleles of the thyroglobulin gene in a Brazilian family with congenital goiter
68 Congdon T, Nguyen LQ, Nogueira CR, Habiby RL, Medeiros-Neto G, and hypothyroidism. Thyroid 1998;8:291–7.
Kopp P. A novel mutation (Q40) in PAX8 associated with congenital 95 Van de Graff SAR, Ris-Stalpers C, Veenboer GJM, Cammenga M, Santos C,
hypothyroidism and thyroid hypoplasia: evidence for phenotypic variability Targovnik HM, de Vijlder JM, Medeiros-Neto G. A premature stop codon in
in mother and child. J Clin Endocrinol Metab 2001;86:3962–7. thyroglobulin messenger RNA results in familial goiter and moderate
69 Meeus L, Gilbert B, Rydlewski C, Parma J, Roussie AL, Abramowicz M, hypothyroidism. J Clin Endocrinol Metab 1999;84:2537–42.
Vilain C, Christophe D, Costagliola S, Vassart G. Characterization of a novel 96 Hishinuma A, Takamatsu J, Ohyama Y, Yokozawa T, Kanno Y, Kuma K,
loss of function mutation of PAX8 in a familial case of congenital Yoshida S, Matsuura N, Ieiro T. Two novel cysteine substitutions (C1263R
hypothyroidism with in-place, normal-sized thyroid. J Clin Endocrinol Metab and C1995S) of thyroglobulin cause a defect in intracellular transport of
2004;89:4285–91. thyroglobulin in patients with congenital goiter and the variant type of
70 Fraizer GC, Shimamura R, Zhang X, Saubders GF. PAX8 regulates human adenomatous goiter. J Clin Endocrinol Metab 1999;84:1438–44.
WT1 transcription through a novel DNA binding site. J Biol Chem 97 Kim PS, Hossain SA, Park Y-N, Lee I, Yoo S-E, Arvan P. A single amino acid
1997;272:30678–87. change in the acetylcholinesterase-like domain of thyroglobulin causes
71 Ohno M, Zannini M, Levy O, Carrasco N, Di Lauro R. The paired-domain congenital goitre with hypothyroidism in the cog/cog mouse: a model of
transcription factor Pax8 binds to the upstream enhancer of the rat sodium/ human endoplasmic reticulum storage diseases. Proc Natl Acad Sci USA
iodide symporter gene and participates in both thyroid-specific and cyclic- 1998;95:9909–13.
AMP-dependent transcription. Mol Cell Biol 1999;19:2051–60. 98 Medeiros-Neto G, Kim PS, Yoo SE, Vono J, Targovnik H, Camargo R,
72 Espinoza CR, Schmitt TL, Loos U. Thyroid transcription factor 1 and Pax8 Hossain SA, Arvan P. Congenital hypothyroid goiter with deficient
synergistically activate the promoter of human thyroglobulin gene. J Mol thyroglobulin. Identification of an endoplasmic reticulum storage disease
Endocrinol 2001;27:59–67. with induction of molecular chaperones. J Clin Invest 1996;98:2838–44.
73 Kozasa T, Itoh H, Tsukamoto T, Kaziro Y. Isolation and characterization of 99 Kim PS, Kwon O-Y, Arvan P. An endoplasmic reticulum storage disease
the human Gsa gene. Proc Natl Acad Sci USA 1988;85:2081–5. causing congenital goiter with hypothyroidism. J Cell Biol
74 Aldred MA, Trembath RC. Activating and inactivating mutations in the 1996;133:517–27.
human GNAS1 gene. Hum Mutation 2000;16:183–9. 100 Dai G, Levy O, Carrasco N. Cloning and characterization of the thyroid
75 Wilson LC, Oude Luttikhuis MEM, Clayton PT, Fraser WD, Trembath RC. iodide transporter. Nature 1996;379:458–60.
Parental origin of Gsa gene mutations in Albright’s hereditary 101 Smanik PA, Ryu K-Y, Theil KS, Mazzaferri EL, Jhiang SN. Expression, exon-
osteodystrophy. J Med Genet 1994;31:835–9. intron organization, and chromosome mapping of the human sodium iodide
76 Yu D, Yu S, Schuster V, Kruse K, Clericuzio CL, Weinstein LS. Identification of symporter. Endocrinology 1997;138:3555–8.
two novel deletion mutations within the Gs alpha gene (GNAS1) in Albright 102 Wolff J. Congenital goiter with defective iodide transport. Endocr Rev
hereditary osteodystrophy. J Clin Endocrinol Metab 1999;84:3254–9. 1983;4:240–54.
77 de Vijlder JJM, Vulsma T. Hereditary Metabolic Disorders causing 103 Fujiwara H, Tatsumi K, Miki K, Harada T, Miyai K, Takai S-I, Amino N.
hypothyroidism. In: Braverman LE, Utiger RD, eds. Werner and Ingbar’s the Congenital hypothyroidism caused by a mutation in the Na+/I2 symporter.
thyroid, 7th ed. Philadelphia: Lippincott-Raven, 1996:749–55. Nat Genet 1997;16:124–5.

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com
Genetics of congenital hypothyroidism 389

104 Kosugi S, Sato Y, Matsuda A, Ohyama Y, Fujieda K, Inomata H, Kameya T, 119 Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S. A novel
Isozaki O, Jhiang SM. High prevalence of T354P sodium/iodide symporter syndrome combining thyroid and neurological abnormalities is associated
gene mutation in Japanese patients with iodide transport defect who have with mutations in a monocarboxylate transporter gene. Am J Hum Genet
heterogeneous clinical pictures. J Clin Endocrinol Metab 1998;83:4123–9. 2004;74:168–75 (Erratum in: Am J Hum Genet 2004;74:598.)
105 Pohlenz J, Duprez L, Weiss RE, Vassart G, Refetoff S, Costagliola S. Failure 120 Agrawal P, Ogilvy-Stuart A, Lees C. Intrauterine diagnosis and
of membrane targeting causes the functional defect of two mutant sodium management of congenital hypothyroidism. Ultrasound Obstet Gynecol
iodide symporters. J Clin Endocrinol Metab 2000;85:2366–9. 2002;19:501–5.
106 Reardon W, Trembath RC. Pendred syndrome. J Med Genet 121 Becks GP, Burrow GN. Diagnosis and treatment of thyroid disease during
1996;33:1037–40. pregnancy. In: DeGroot LJ, Besser M, Burger HG, Jameson JL, Loriaux DL,
107 Mondini C. Anatomia surdi nati sectio: de Boroniensi scientarium et artium Marshall JC, Odell WD Potts JT, Rubenstein AAAH, eds. Endocrinology, 3rd
instituto atque acedemia commentarii. Bononiae 1791;7:419–31. ed. Philadelphia: WB Saunders, 1995:799–820.
108 Reardon W, O’Mahoney CF, Trembath R, Jan H, Phelps PD. Enlarged 122 Kohn LD, Harii N. Thyrotropin receptor autoantibodies (TSHRAbs): epitopes,
vestibular aqueduct: a radiological marker of Pendred syndrome, and origins and clinical significance. Autoimmunity 2003;36:331–7.
mutation of the PDS gene. Q J Med 2000;93:99–104. 123 Pohlenz J, Rosenthal IM, Weiss RE, Jhiang SM, Burant C, Refetoff S.
109 Batsakis JG, Nishiyama RH. Deafness with sporadic goitre. Arch Congenital hypothyroidism due to mutations in the sodium/iodide
Otolaryngol 1962;76:401–6. symporter. J Clin Invest 1998;101:1028–35.
110 Masmoudi S, Charfedine L, Hmani M, Grati M, Ghorbel AM, Elgaied- 124 Bongers-Schokking JJ, Koot HM, Wiersma D, Verkerk PH, de Mulnck
Boulila A, Drira M, Hardelin J-P, Ayadi H. Pendred syndrome: phenotypic Keizer-Shrama SMPF. Influence of timing and dose of thyroid hormone
variability in two families carrying the same PDS missense mutation. replacement on development in infants with congenital hypothyroidism.
Am J Med Genet 2000;90:38–44. J Pediatr 2000;136:292–7.
111 Everett LA, Glaser B, Beck JC, Idol JR, Buchs A, Heyman M, Adawi F, 125 Derksen-Lubsen G, Verkerk PH. Neuropsychologic development in early
Hazani E, Nassir E, Baxevanis AD, Sheffield VC, Green ED. Pendred treated congenital hypothyroidism: analysis of literature data. Pediatr Res
syndrome is caused by mutations in a putative sulphate transporter gene 1996;39:561–6.
(PDS). Nat Genet 1997;17:411–22.
126 Dubuis JM, Glorieux J, Richer F, Deal CL, Dussault JH, Van Vliet G. Outcome
112 Royaux IE, Suzuki K, Mori A, Katoh R, Everett LA, Kohn LD, Green ED.
of severe congenital hypothyroidism: closing the developmental gap with
Pendrin, the protein encoded by the Pendred syndrome gene (PDS), is an
early high dose levothyroxine treatment. J Clin Endocrinol Metab
apical porter of iodide in the thyroid and is regulated by thyroglobulin in
1996;81:222–7.
FRTL-5 cells. Endocrinology 2000;14:839–45.
127 Heyerdahl S, Kase BF, Lie SO. Intellectual development in children with
113 Scott DA, Wang R, Kreman TM, Andrews M, McDonald JM, Bishop JR,
congenital hypothyroidism in relation to recommended thyroxine treatment.
Smith RJH, Karniski LP, Sheffield VC. Functional differences of the PDS gene
products are associated with phenotypic variation in patients with Pendred J Pediatr 1991;18:850–7.
syndrome and non-syndromic hearing loss (DFNB4). Hum Mol Genet 128 Tillotson SL, Fuggle PW, Smith I, Ades AE, Grant DB. Relation between
2000;9:1709–15. biochemical severity and intelligence in early treated congenital
114 Caillou B, Dupuy C, Lacroix L, Nocera M, Talbot M, Ohayon R, Deme D, hypothyroidism: a threshold effect. BMJ 1994;309:440–5.
Bidart J-M, Schlumberger M, Virion A. Expression of reduced nicotinamide 129 Manley NR, Capecchi MR. The role of Hoxa-3 in mouse thymus and thyroid
adenine dinucleotide phosphate oxidase (ThoX, LNOX, Duox) genes and development. Development 1995;121:1989–2003.
proteins in human thyroid tissues. J Clin Endocrinol Metab 2001;86:3351–8. 130 de la Chapelle A, Herva R, Koivisto M, Aula P. A deletion in chromosome 22
115 Corvilain B, Van Sande J, Laurent E, Dumont JE. The H2O2-generating can cause DiGeorge syndrome. Hum Genet 1981;57:253–6.
system modulates protein iodination and the activity of the pentose 131 Pueblitz S, Weinberg AG, Albores-Saavedra J. Thyroid C cells in the
phosphate pathway in dog thyroid. Endocrinology 1991;128:779–85. DiGeorge anomaly: a quantitative study. Pediatr Pathol
116 Pachucki J, Wang D, Christophe D, Miot F. Structural and functional 1993;13:463–73.
characterization of two human ThOX/Duox genes and their 59 flanking 132 Meunier D, Aubin J, Jeannotte L. Perturbed thyroid morphology and
regions. Mol Cell Endocrinol 2004;214:53–62. transient hypothyroidism symptoms in Hoxa5 mutant mice. Dev Dyn
117 Moreno JC, Bikker H, Kempers MJ, van Trotsenburg AS, Baas F, de Vijlder JJ, 2003;227:367–78.
Vulsma T, Ris-Stalpers C. Inactivating mutations in the gene for thyroid 133 Lints TJ, Parsons LM, Hartley L, Lyons I, Harvey RP. Nkx-2. 5: a novel murine
oxidase 2 (THOX2) and congenital hypothyroidism. N Engl J Med homeobox gene expressed in early heart progenitor cells and their myogenic
2002;347:95–102. descendants, Development 1993;119:419–31.
118 Friesema EC, Ganguly S, Abdalla A, Manning Fox JE, Halestrap AP, 134 Roberts HE, Moore CA, Fernhoff PM, Brown AL, Khoury MJ. Population
Visser TJ. Identification of monocarboxylate transporter 8 as a specific study of congenital hypothyroidism and associated birth defects, Atlanta,
thyroid hormone transporter. J Biol Chem 2003;278:40128–35. 1979–1992. Am J Med Genet 1997;71:29–32.

www.jmedgenet.com
Downloaded from jmg.bmj.com on May 17, 2012 - Published by group.bmj.com

Genetics of congenital hypothyroidism


S M Park and V K K Chatterjee

J Med Genet 2005 42: 379-389


doi: 10.1136/jmg.2004.024158

Updated information and services can be found at:


http://jmg.bmj.com/content/42/5/379.full.html

These include:
References This article cites 128 articles, 63 of which can be accessed free at:
http://jmg.bmj.com/content/42/5/379.full.html#ref-list-1

Article cited in:


http://jmg.bmj.com/content/42/5/379.full.html#related-urls

Email alerting Receive free email alerts when new articles cite this article. Sign up in the
service box at the top right corner of the online article.

Notes

To request permissions go to:


http://group.bmj.com/group/rights-licensing/permissions

To order reprints go to:


http://journals.bmj.com/cgi/reprintform

To subscribe to BMJ go to:


http://group.bmj.com/subscribe/

You might also like